Acute Bacterial Meningitis in Infants and Children

March 27, 2018 | Author: Maha Stephania Hafez | Category: Meningitis, Antimicrobial Resistance, Streptococcus, Antibiotics, Cerebrospinal Fluid


Comments



Description

ReviewAcute bacterial meningitis in infants and children Kwang Sik Kim Lancet Infect Dis 2010; 10: 32–42 Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA (Prof K S Kim MD) Correspondence to: Prof Kwang Sik Kim, Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, 200 North Wolfe Street, Room 3157, Baltimore, MD 21287, USA [email protected] Bacterial meningitis continues to be an important cause of mortality and morbidity in neonates and children throughout the world. The introduction of the protein conjugate vaccines against Haemophilus influenzae type b, Streptococcus pneumoniae, and Neisseria meningitidis has changed the epidemiology of bacterial meningitis. Suspected bacterial meningitis is a medical emergency and needs empirical antimicrobial treatment without delay, but recognition of pathogens with increasing resistance to antimicrobial drugs is an important factor in the selection of empirical antimicrobial regimens. At present, strategies to prevent and treat bacterial meningitis are compromised by incomplete understanding of the pathogenesis. Further research on meningitis pathogenesis is thus needed. This Review summarises information on the epidemiology, pathogenesis, new diagnostic methods, empirical antimicrobial regimens, and adjunctive treatment of acute bacterial meningitis in infants and children. Introduction Bacterial meningitis, an inflammation of the meninges affecting the pia, arachnoid, and subarachnoid space that happens in response to bacteria and bacterial products, continues to be an important cause of mortality and morbidity in neonates and children.1–4 However, mortality and morbidity vary by age and geographical location of the patient and the causative organism. Patients at risk for high mortality and morbidity include newborns, those living in low-income countries, and those infected with Gram-negative bacilli and Streptococcus pneumoniae.1–4 Severity of illness on presentation (eg, low score on Glasgow coma scale), infection with antimicrobialresistant organisms, and incomplete knowledge of the pathogenesis of meningitis are additional factors contributing to mortality and morbidity associated with bacterial meningitis.1–7 Suspected bacterial meningitis is a medical emergency; thus, immediate steps must be taken to establish the specific diagnosis, and empirical antimicrobial treatment must be started rapidly. The mortality of untreated bacterial meningitis approaches 100% and, even with optimum treatment, mortality and morbidity might happen. Neurological sequelae are relatively common in survivors of meningitis, particularly after pneumococcal meningitis.1–6 Epidemiology Almost all microbes that are pathogenic to human beings have the potential to cause meningitis, but a relatively small number of pathogens (ie, group B streptococcus, Escherichia coli, Listeria monocytogenes, Haemophilus influenzae type b [Hib], S pneumoniae, and Neisseria meningitidis) account for most cases of acute bacterial meningitis in neonates and children, although the reasons for this association remain incompletely understood. The absence of an opsonic or bactericidal antibody is a major risk factor in most cases of meningitis caused by group B streptococcus, E coli, Hib, S pneumoniae, and N meningitidis.8–12 Age-related incidence of Hib and N meningitidis disease is inversely related to prevalence of serum bactericidal activity,8,10 and the lack of type-specific 32 antibody is a major risk factor for neonatal group B streptococcal disease.11 Determinations of microbial targets capable of inducing opsonic or bactericidal antibodies and successful vaccination programmes with such targets in infants and children have changed the epidemiology of bacterial meningitis.13–18 However, microbial targets for opsonic or bactericidal antibodies have not been determined against all pathogens that commonly cause meningitis. The advancement of vaccine design in enhancing immunogenicity has been shown to be important in preventing meningitis caused by Hib, S pneumoniae, and N meningitidis. Protein-conjugated capsular polysaccharide vaccines have almost completely eliminated meningitis caused by vaccine serotypes. Routine immunisation in young infants and children with Hib conjugate vaccines has virtually eradicated meningitis due to these organisms in many high-income countries;13 in the USA, Hib meningitis happens primarily in children that are not immunised and among infants too young to have completed the primary immunisation series.14 Additionally, introduction of the seven-valent pneumococcal conjugate vaccine (PCV7) has led to a substantial reduction in the incidence of pneumococcal meningitis in infants and children younger than 5 years.15–17 Use of these protein-conjugated vaccines has also reduced Hib and pneumococcal meningitis among unvaccinated populations through herd immunity. At present, limitations with PCV7 and meningococcal conjugate vaccines include an apparent increase in the incidence of invasive pneumococcal disease, including meningitis caused by non-PCV7 serotypes, such as serotype 19A (a penicillin and third-generation cephalosporin-resistant non-PCV7 serotype), and an apparent decline in bactericidal antibody against N meningitidis in infants, requiring a booster immunisation in the second year of life.17,18 Pathogenesis A relatively small number of microbial pathogens has been shown to account for most cases of meningitis in infants and children, but how those pathogens cross the blood–brain barrier and cause meningitis is incompletely www.thelancet.com/infection Vol 10 January 2010 40 but its contribution to Hib traversal of the blood–brain barrier is unclear.7. adeno-associated virus. pili. group B streptococcus.60.7. but the mature monomer is shown to be present on the cell surface and functions as a membrane receptor for the adhesive basement membrane protein laminin. as shown by partial inhibition of pneumococcal invasion of HBMEC by a PAFR antagonist.7.41 L monocytogenes penetration into the CNS has been attributed to transmigration of L monocytogenes-infected monocytes and myeloid cells across the blood–brain barrier.Review Bacterium Blood Brain endothelial cell Binding Brain Invasion Traversal Figure: Bacterial interaction with the blood–brain barrier.53 The monomer of RPSA (37 kDa laminin receptor protein) is a ribosomeassociated cytoplasmic protein and a precursor of the 67 kDa laminin receptor. it acts as a cellular receptor for L monocytogenes Vip.31 However. affecting different host signalling molecules.54 RPSA has also been shown to be a cellular target for various CNS-infecting microorganisms (table 1). contributing to penetration into the brain understood. hepatocyte growth factor.19 Transcellular traversal of the blood–brain barrier has been shown for most meningitis-causing pathogens in infants and children.7.thelancet.61 although the main route of L monocytogenes penetration into the CNS still needs to be determined.30. which include the receptor for the globular head of complement component C1q (gC1q-R) and Met tyrosine kinase. which is involved in infection of the spleen.30. www.57. For example.31 E coli invasion of HBMEC has also been shown to happen through other interactions with host receptors.33–37 The mechanism by which the same receptor is involved in CNS penetration by different organisms remains to be established. such as group B streptococcus and L monocytogenes.30 In addition.22.25–29 Meningitis-causing pathogens cross the blood–brain barrier transcellularly. Venezuelan equine encephalitis virus.62 PAFR has also been shown to interact with Hib (table 1).19 The E coli proteins that contribute to HBMEC binding (ie. liver.56 but whether these structures are unique to meningitis isolates of group B streptococcus is unclear.42 Several HBMEC receptors for InlB have been identified. group B streptococcus. InlB does not compete for the same interaction site on Met tyrosine kinase as the natural ligand.38. N meningitidis invasion of HBMEC is mediated by the outer membrane protein Opc binding to fibronectin. dengue virus. N meningitidis.25–29 For example. and brain of mice. and N meningitidis. However.32. paracellularly. and S pneumoniae (figure). Hib. 33 . CD48 and endoplasmin (formerly gp96).19.19. FbsA (fibrinogen-binding protein). and IagA (via lipoteichoic acid anchoring). FimH and OmpA) do so through interactions with their respective HBMEC receptors.20–23 The blood–brain barrier is a structural and functional barrier that is formed by brain microvascular endothelial cells.24 which protects the brain from any microbes and toxins circulating in the blood.58 but their contributions to L monocytogenes invasion of HBMEC remain incompletely understood. L monocytogenes invasion of HBMEC is mediated by internalin B (InlB).7.55.19 Experimental animal models and human cases of meningitis suggest that E coli and group B streptococcus penetrate the brain initially through the cerebral vasculature. possess several microbial structures that allow their binding to and invasion of HBMEC. including E coli. meningitis-causing pathogens.19. including E coli. including S pneumoniae. E coli penetration into the brain involves its binding to and invasion of the human brain microvascular endothelial cells (HBMEC) that constitute the blood–brain barrier.25–28 Recent studies have shown that microbial traversal of the blood–brain barrier happens via microbial interactions with host receptors (table 1).28.47–49 Endoplasmin is an endoplasmic reticulum paralogue of heat shock protein 90 that is also present on the surface of HBMEC.25.com/infection Vol 10 January 2010 endoplasmin also interacts with OmpA.50–52 For example. and prion protein. S pneumoniae.19.7. have been shown to cross the blood–brain barrier as live bacteria.39 and delayed translocation of pneumococci from the lung to the blood and from the blood to the cerebrospinal fluid (CSF) in PAFR-knockout mice. Other meningitis-causing pathogens. It is unclear how the laminin receptor is matured and synthesised from the laminin receptor protein.59 gC1q-R is also the HBMEC receptor for Plasmodium falciparum-infected erythrocytes (table 1). or by means of infected phagocytes (so-called Trojan horse mechanism). cytotoxic necrotising factor 1 (CNF1) interacts with 40S ribosomal protein subunit A (RPSA) on HBMEC. Group B streptococcal binding to HBMEC happens via Lmb (laminin-binding protein). S pneumoniae crosses the blood–brain barrier partly through interaction between cell-wall phosphorylcholine and the platelet-activating factor receptor (PAFR). and respiratory depression (Cushing’s triad) is a late sign of increased intracranial pressure.63 CD46 has also shown to be a receptor for measles. interleukin-8 secretion in response to E coli strain K1 happens in HBMEC. the symptoms and signs depend on the age of the child.70 Similar findings were seen for a group B streptococcus Lmb mutant. clinical features might include fever. lethargy. they might include fever. Hib=Haemophilus influenzae type b.Review Ligands References Endoplasmin Escherichia coli OmpA 30 Listeria monocytogenes Vip 31 37 kDa laminin receptor protein Escherichia coli CNF1 32 Neisseria meningitidis PilQ/PorA 33 Streptococcus pneumoniae CbpA 33 Hib Omp2 33 Prion protein ·· 34 Viruses (sindbis. 30% had bacterial meningitis.43 and lipo-oligosaccharides have been shown to contribute to a high-degree of bacteraemia and subsequent penetration into the CNS.71 In older children.19. Of note. For example. tick-borne encephalitis.38 In addition. or even absent. However. the mechanisms involved in microbial invasion of the blood–brain barrier differ from those involved in the release of cytokines and chemokines in response to meningitis-causing pathogens. In infants. nausea. bradycardia. hypothermia. although the release of interleukins 6 and 8 from HBMEC in response to bacterial invasion involves the p38 mitogen-activated protein kinase pathway. photophobia. Signs of meningeal irritation are present in 75% of children with bacterial meningitis at the time of presentation. Venezuelan equine encephalitis.53. and if there is no cardiopulmonary compromise.44–46 The involvement of host receptors and signaltransduction pathways in the microbial invasion of the blood–brain barrier might provide a new way to prevent and treat meningitis by the targeting of such host receptors or signalling molecules. and increased intracranial pressure. cytosolic phospholipase A2α) were efficient in preventing E coli penetration into the brain. confusion.19.64 and could indeed be used to prevent or treat meningitis.64–69 A proof-ofconcept study has shown that down-modulation of the HBMEC receptor for CNF1 (RPSA) and blockade or inhibition of host molecules involved in E coli invasion of HBMEC (eg. but not in non-brain endothelial cells (eg. In a study of neonatal meningitis.64 Recent studies suggest that this concept is also relevant to other meningitis-causing pathogens. respiratory distress. vomiting. seizures. or irritability. in a retrospective review of 326 children presenting to a paediatric emergency department in the Netherlands between 1988 and 1998 with signs of meningeal irritation. human 34 umbilical vein endothelial cells). variable.33. fever or hypothermia was noted in 62% of cases. E coli proteins involved in binding to and invasion of HBMEC did not affect the release of interleukin 8 from HBMEC. lethargy.73 Absence of meningeal irritation in children with bacterial meningitis was substantially more common in those younger than 12 months. Brudzinski’s sign (passive flexion of the neck elicits flexion of the hips). non-specific. adeno-associated) ·· 35–38 Streptococcus pneumoniae Phosphorylcholine 39 Hib Phosphorylcholine 40 Plasmodium falciparum Infected erythrocytes 41 Listeria monocytogenes InlB 42 Platelet-activating factor receptor gC1q-R CD46 Neisseria meningitidis Pili 43 Measles Haemagglutinin 44 Adenovirus Ad35 knob 45 Human herpesvirus 6 Glycoprotein H 46 CNF1=cytotoxic necrotising factor 1. and human herpesvirus 6 (table 1). or bulging fontanelles. focal neurological findings.com/infection Vol 10 January 2010 . However. but induced equal concentrations of interleukin 8 compared with the parent strain. poor feeding. Other signs of bacterial meningitis on physical examination include Kernig’s sign (flexing the hip and extending the knee to elicit pain in the back and legs).74 The constellation of systemic hypertension. and the host response to infection.7. Table 1: Blood–brain barrier receptors used by CNS-infecting microorganisms thereby anchoring the bacteria to the integrin α5β1 receptor on the cell surface. N meningitidis pili bind to CD46 on HBMEC.19. The clinical features of bacterial meningitis in infants and children can be subtle. N meningitidis invasion of HBMEC has been shown to involve c-Jun kinases 1 and 2.29 In addition. adenovirus. Evidence for mass www. Patients with suspected meningitis should receive a lumbar puncture after a mass lesion has been ruled out on clinical grounds or by CT scan of the head. diarrhoea.32. Diagnosis Clinical findings Bacterial meningitis requires early diagnosis and empirical antimicrobial treatment. which was defective for the invasion of HBMEC. irritability.thelancet. the duration of illness. vomiting. dengue. Laboratory findings CSF examination is of paramount importance for the diagnosis of all forms of meningitis (table 2). headaches.68 These findings suggest that targets for prevention of bacterial penetration into the brain differ from those involved in CNS inflammation associated with meningitis. gC1q-R=receptor for the globular head of complement component C1q.72 By contrast. and in four of 14 CSF samples that were culture-negative. 16 copies of L monocytogenes.80 In such children. In the presence of homologous antigen there is visible agglutination of the antibody-coated latex beads. and S pneumoniae. latex agglutination was positive in 49 (66%) of 74 CSF samples that grew S pneumoniae. In addition.82 whereas the sensitivity for broad-range 16S ribosomal DNA PCR was about 10–200 organisms per mL CSF. Real-time PCR has been shown to detect as few as two copies of E coli. Escherichia coli. microarray or biochip.81 In the multicentre pneumococcal meningitis surveillance study. PCR. and concentrations of protein and glucose are helpful in the differential diagnosis of various forms of meningitis (table 2). and a third of patients with listeria meningitis. Latex agglutination uses latex beads adsorbed with microbe-specific antibodies.6 CSF culture can be negative in children who receive antibiotic treatment before CSF examination.Review lesions will include focal neurological signs and evidence of increased intracranial pressure. and Haemophilus influenzae type b.com/infection Vol 10 January 2010 Opening pressure (cm H20) White blood cells (×10⁶ cells per L) Glucose (mg/dL) Protein (mg/dL) Common >20 >1000 <10 >100 Less common <20 Bacteria* 5–1000 10–45 50–100 Mycobacterium tuberculosis Common >20 100–500 Less common <20 5–100 Common <20 100–500 Less common <20 5–100 10–45 <10 >100 50–100 Borrelia burgdorferi 10–45 <10 50–150 >150 Treponema pallidum Common <20 Less common <20 5–500 >500 10–45 <10 50–150 >150 Fungi Common Variable Less common Variable 5–500 >500 10–45 <10 >100 50–100 Viruses Common <20 Less common <20 5–500 >500 Normal 10–45 50–100 >100 *Group B streptococci. which amplifies DNA under isothermal conditions (63°C). half of patients with Gramnegative bacillary meningitis. Table 2: Likely pathogens for CNS infections on the basis of cerebrospinal fluid analysis previously received antibiotics.thelancet. is a promising tool. loop-mediated isothermal amplification method. increased CSF white blood cell counts and increased CSF protein concentration are usually sufficient to establish the diagnosis of bacterial meningitis. because it does not need thermocycling apparatus and the results can be read with the naked eye (based on turbidity or colour development by SYBR Green dye for 35 . complete sterilisation of N meningitidis from CSF happened within 2 h of giving a parenteral thirdgeneration cephalosporin and the beginning of sterilisation of S pneumoniae from CSF by 4 h into treatment. Listeria monocytogenes.6 The use of standard or sequential-multiplex PCR has been shown to be useful in identification of infecting pathogens in patients who have previously received antibiotics or in resource-poor settings. the limit of detection differs between assays. Such tests include latex agglutination. but less sensitive with N meningitidis antigen.86 which might provide more rapid and accurate diagnosis of bacterial infection in infants and children. or whose initial CSF Gram stain is negative with negative culture at 72 h incubation.85 The time needed for the whole process from DNA extraction to the end of real-time PCR was 1·5 h.75–78 Gram stain is positive in about 90% of children with pneumococcal meningitis.78. Streptococcus pneumoniae. Neisseria meningitidis. A Gram stain of CSF will show whether bacteria are present. and immunochromatography (table 3). Non-culture methods Non-culture tests should be considered for patients who need earlier identification of pathogens or have previously received antibiotics.79 CSF cell count and differential. N meningitidis.82–87 Multiplex realtime PCR or broad-range PCR aimed at the 16S ribosomal RNA gene of eubacteria is promising for the detection of pathogens from CSF. Blood cultures or non-culture diagnostic tests might help in identifying the infecting pathogen. particularly in resource-poor settings. A low CSF white blood cell count with positive Gram stain is a risk factor for an unfavourable outcome.87 A recently developed nucleic-acid amplification technique. and a positive Gram stain shows bacterial counts higher than 1×10³ cells per mL in CSF.82 an attractive timeframe for its application in clinical practice. and 28 copies of group B streptococcus. The detection rate was substantially higher with PCR than with cultures in patients who had www. A Gram-stain-specific probe-based real-time PCR using 16S ribosomal RNA has been shown to allow simultaneous detection and discrimination of clinically relevant Grampositive and Gram-negative bacteria directly from blood samples.84. loop-mediated isothermal amplification. For example. about 80% of children with meningococcal meningitis. sequential PCR-based serotyping of S pneumoniae using serotype-specific primers could improve ascertainment of pneumococcal serotype distribution in settings in which prior use of antibiotics is high. Latex agglutination assays have been sensitive towards Hib antigen. and CSF pressure should be recorded during the lumbar puncture.75–78 Cytospin centrifugation increases the chances of detecting organisms in Gramstained CSF.82 However. and bactericidal antibiotics should be administered intravenously at the highest clinically validated doses to patients with suspected bacterial meningitis.7 36 Antimicrobial treatment Eradication of the infecting organism from the CSF is entirely dependent on antibiotics. empirical treatment for patients with bacterial meningitis in areas where resistant S pneumoniae strains are prevalent must include the addition of vancomycin (panel).95 Because bacterial meningitis is defined as inflammation that happens in response to bacteria and bacterial products. and 5·5% of meningitis cases happened without pleocytosis. blood neutrophil count of at least 10×10⁹ cells per L.105 Therefore. β-lactams and vancomycin) have decreased penetration into CSF in the absence of meningeal inflammation.90–92 However. whereas a regimen for Gram-negative bacilli with a high likelihood of resistance (eg. For example.88. Selection of empirical antimicrobial regimens is designed to cover the likely pathogens. this proposed diagnostic tool only achieved 95% sensitivity. with modifications if CSF Gram stain is positive. gentamicin).95 For example.94. whereas hydrophilic agents (ie. five patients with bacterial meningitis who had pleocytosis were found to have a bacterial meningitis score that indicated low risk.93 Compared with CSF culture (sensitivity of 71%) and latex agglutination (86%). and hybridisation of labelled DNA with oliogonucleotide probes (pathogenspecific or virulence genes) immobilised on a microarray. potentially useful in resource-poor settings Microarray or biochip90–92 Not yet Requires a suitable biochip Immunochromatography93 Not yet Highly sensitive for Streptococcus pneumoniae Table 3: Non-culture diagnostic tests for identification of pathogens for meningitis staining nucleic acids).102–104 An important factor in the choice of empirical antimicrobial agents is the emergence of antimicrobialresistant organisms. fluoroquinolones and rifampicin) penetrate relatively well into the CSF even if the meninges are not inflamed. and proteinbinding ability of drugs.102. patients with CSF culture positivity without pleocytosis or increased CSF protein concentrations are presumably representative of the early stages of bacterial meningitis. amplification of targeted DNA. and outpatient management might be considered for children who had pleocytosis (7×10⁶ cells per L or more) and none of the following five criteria on presentation: history of a seizure with the illness. molecular weight. CSF protein of at least 80 mg/dL.97 Several retrospective and prospective studies showed that delay in antibiotic treatment was associated with adverse outcomes. www. and many of the penicillin-resistant pneumococci have reduced susceptibility to third-generation cephalosporins (ie. and Gram-negative bacilli that are resistant to many β-lactam drugs. and efflux transporters.96. inflammation of the meninges. the prevalence of S pneumoniae strains that are relatively resistant to penicillin (minimum inhibitory concentration [MIC] 0·1–1·0 μg/mL) or highly resistant to penicillin (MIC greater than 1·0 μg/mL) is increasing. or CSF neutrophil count of at least 1×10⁹ cells per L. The ability of an antimicrobial agent to penetrate the blood–brain barrier is the most important factor that determines whether efficient bacterial killing happens in the CSF. including S pneumoniae that is resistant to penicillin or third-generation cephalosporins. penetration of vancomycin into the CSF can be reduced in the absence of meningeal inflammation and also in patients who receive adjunctive dexamethasone treatment.98–101 In patients with suspected bacterial meningitis for whom immediate lumbar puncture is delayed due to pending brain imaging study or the presence of disseminated intravascular coagulation. A rapid immunochromatographic test for S pneumoniae was evaluated in 122 children with pneumococcal meningitis. Bacterial meningitis score The ability to distinguish between bacterial and nonbacterial aseptic meningitis in infants and children in the emergency department could contribute to limiting hospital admissions or unnecessary use of antibiotics.97 Treatment failures in bacterial meningitis as a result of multiresistant organisms have been reported. suggesting that immunochromatography might be useful in the diagnosis of pneumococcal meningitis. Treatment of patients at risk of infection with L monocytogenes must include a synergistic regimen containing ampicillin and an aminoglycoside (eg. However.com/infection Vol 10 January 2010 .88 but its use in the diagnosis of bacterial meningitis has not been tested.103 Lipophilic agents (ie. cefotaxime and ceftriaxone). Identification of pathogens by use of a microarray or biochip involves extraction of genomic DNA from CSF.thelancet. Blood–brain-barrier penetration is affected by lipophilic property. its usefulness in clinical practice has not been shown.96. based on age of the patient and specific risk factors (table 4).81 Yes Sensitive with Haemophilus influenzae type b. However.Review Clinical application Comments Latex agglutination78. but less sensitive with Neisseria meningitidis PCR82–87 Not yet Need to develop specific and broad targets or primers Loop-mediated isothermal amplification88. blood cultures must be obtained and antimicrobial treatment should be initiated immediately. positive CSF Gram stain.89 The assay detected ten or more copies of S pneumoniae in oral mucosa swab samples. nosocomial meningitis) should include an aminoglycoside (eg.89 Not yet Does not require thermocycling apparatus. immunochromatography was 100% sensitive for the diagnosis of pneumococcal meningitis. The bacterial meningitis score has been developed for assessing infants and children with meningitis. 115 These findings support the use of a thirdgeneration cephalosporin for meningococcal meningitis in areas where penicillin resistance is prevalent. and thus cannot be used as monotherapy for bacterial meningitis. and has been shown to be uniformly susceptible to β-lactam antibiotics (eg. studies have reported isolates of group B streptococcus with penicillin MICs of 0·12–1·0 μg/mL that had mutations in the target penicillin-binding proteins similar to the mechanisms involved in penicillin-resistant S pneumoniae. gatifloxacin was as effective as the combination of ceftriaxone and vancomycin against a highly cephalosporin-resistant pneumococcal strain in an experimental meningitis model. gatifloxacin. including penicillin against group B streptococcus. and continued monitoring of antimicrobial susceptibility patterns. immunodeficiency.108 However.thelancet. including cerebral palsy and mental retardation. relative resistance to penicillin (MIC 0·1 μg/mL) has been shown to occur in 3–4% of the meningococcal isolates in the USA and in 2% of the 137 isolates recovered between 2000 and 2006 from equatorial sub-Saharan Africa (the so-called meningitis belt). Hib 3–6 months No immunisation S pneumoniae.113 By contrast. Table 4: Likely pathogens for meningitis based on age and immunisation status garenoxacin).97.127 37 . For example.116–124 For example.106 Antibacterial killing activity in CSF also depends on the bacterial burden at the start of treatment.96. diabetes mellitus (S pneumoniae. Similarly.120 Moxifloxacin and garenoxacin had CSF bacterial killing rates that exceeded those found with the combination of ceftriaxone and vancomycin against experimental meningitis caused by vancomycin-tolerant S pneumoniae. MICs of β-lactam antibiotics. For example. group B streptococcus is commonly responsible for neonatal bacterial meningitis. and developmental delay. terminal complement deficiencies. is thus important. However. The penetration of intravenously given aminoglycosides into the CSF remains variable or poor even in the presence of meningeal inflammation. freshmen living in dormitories.114. ertapenem). cochlear implant.102. and thus penicillin is at present the drug of choice for invasive group B streptococcal infection including meningitis. and include hearing loss. caused by group B streptococcus and S pneumoniae) who have many organisms on CSF Gram stain are likely to yield 10⁷–10⁸ organisms per mL. otitis. some patients with bacterial meningitis (eg. The MIC and minimum bactericidal concentration are established in laboratories by use of bacterial inoculum size of 10⁴–10⁵ organisms per mL.126 Hearing loss happens in 22–30% of survivors of pneumococcal meningitis compared to 1–8% after meningococcal meningitis. including newer agents. Listeria monocytogenes). Listeria monocytogenes (neonatal pathogens) 1–3 months No immunisation or one dose of primary immunisation Neonatal pathogens. www. Hib S pneumoniae (non-PCV serotypes).106. but they might be useful if other drugs cannot be used.109. N meningitidis.112 A recent study in Spain reported an increased incidence in penicillin non-susceptible strains of N meningitidis (eg. outbreaks (Neisseria meningitidis). N meningitidis Risk factors for specific pathogens are as follows: cerebrospinal fluid leak. penicillin MIC 0·1 μg/mL or less). sinusitis (S pneumoniae.125 Adjunctive treatment Neurological sequelae are common in survivors of meningitis. gemifloxacin. cognitive impairment.121 Fluoroquinolones are not recommended for use in children younger than 18 years because of concerns about their effects on growing cartilage in experimental animals. HIV infection. The potential roles of newer β-lactam antibiotics (meropenem. S pneumoniae. penicillin has been the standard treatment for meningococcal meningitis. and lipopeptides (daptomycin) in the treatment of meningitis caused by resistant bacteria have been shown in animal models of experimental meningitis.110 The optimum empirical regimen for meningitis caused by penicillin non-susceptible group B streptococci that includes third-generation cephalosporins has not been established. Haemophilus influenzae type b [Hib]). recently developed quinolones (moxifloxacin.111. clinical effectiveness of these newer antimicrobial drugs as monotherapy in the treatment of meningitis caused by penicillin nonsusceptible isolates of S pneumoniae has not been established. dexamethasone did not substantially affect the penetration of gemifloxacin and moxifloxacin into the CSF. nephrotic syndrome (Streptococcus pneumoniae). Antimicrobial susceptibility patterns must be established for all organisms isolated from the CSF. MICs 0·1–0·5 μg/mL) from 9·1% in 1986 to 71·4% in 1997. were increased 1000 times when the inoculum size increased from 10⁴ to 10⁸ organisms per mL. Of interest.6.107 Careful monitoring of the response to antimicrobial treatment is therefore warranted for patients with bacterial meningitis who have high bacterial burden on the basis of initial CSF Gram stain.Review amikacin) plus a third-generation or fourth-generation cephalosporin. N meningitidis >7 months to 5 years No immunisation Primary immunisation completed 6–21 years S pneumoniae.76 and MIC values can be 100–1000-times higher than would normally be expected. Escherichia coli. For example. PCV=pneumococcal conjugate vaccine. Hib At least two doses of primary immunisation (with Hib-Omp vaccine) S pneumoniae. with an implication of treatment failures. sickle-cell disease.124 However. N meningitidis. but penicillin resistance has evolved. at least until penicillin susceptibility is known.119. or meropenem. N meningitidis S pneumoniae. N meningitidis.6. asplenia. the Metropolitan Atlanta Developmental Disabilities Surveillance Program in 1991 identified bacterial meningitis as the leading postnatal cause of developmental disabilities. cefepime.com/infection Vol 10 January 2010 Likely pathogens <1 month Group B streptococci. including hippocampal apoptosis in experimental animals with pneumococcal and E coli meningitis who received dexamethasone. positive Gram stain) 3 months to 21 years Cefotaxime (75 mg/kg every 6–8 h.132 Another issue with adjunctive dexamethasone treatment is the possibility of neuronal injury. Success with the protein-conjugate Hib and S pneumococcus PCV vaccines in the prevention of meningitis shows that identification of conserved targets for opsonic or bactericidal antibodies is likely to enhance the development of effective vaccination programmes for the prevention of meningitis caused by N meningitidis and other meningitis-causing bacteria. particularly for patients with meningitis caused by pneumococci that are resistant to third-generation antibiotics. up to a maximum of 600 mg daily) can be added in the setting of administration of dexamethasone In a 2007 Cochrane review. secondary fever (recurrence of fever after at least 24 h without fever) happens more commonly in patients treated with dexamethasone than in those who are not (52% vs 24%. Another important consideration for the treatment of bacterial meningitis is the substantial morbidity in survivors of meningitis. individuals living in low-income countries. Dexamethasone given shortly before or when antibiotics were first given has been shown to reduce the rate of hearing loss in children with Hib meningitis. by age Less than 1 month Ampicillin (50–100 mg/kg every 6 h) plus gentamicin (2·5 mg/kg every 8 h). and such cases should be treated with dexamethasone as well as vancomycin and ceftriaxone. Most meningitiscausing pathogens cross the blood–brain barrier. fever curve) can be complicated by the use of dexamethasone.134 Long-term follow-up studies are thus needed to address the effect of dexamethasone treatment on any cognitive and neuropsychological outcomes in patients with bacterial meningitis. in whom bacteriological killing in the CSF depends on vancomycin. multi-resistant Gram-negative bacilli). its use as adjunctive treatment in children with bacterial meningitis.6 In addition. is promising. Emergence of antimicrobial-resistant bacteria presents a constant challenge to the development of new bactericidal antibiotics for the treatment of bacterial meningitis.130 The widespread use of dexamethasone in children with bacterial meningitis needs careful monitoring of clinical (eg.129 The American Academy of Pediatrics Committee on Infectious Diseases suggests that dexamethasone treatment might be considered for infants and children older than 6 weeks with pneumococcal meningitis after considering the potential benefits and possible risks.6. and infections caused by antimicrobial-resistant pathogens (eg. Advances in microbial genome sequencing and functional genomic approaches are likely to be beneficial in the identification of such microbial targets. or rifampicin (10 mg/kg every 12 h. but its beneficial effects on hearing and other neurological sequelae are not as clear against meningitis caused by other organisms. up to a maximum 1 g per dose).thelancet. p=0·0009). 95% CI 0·31–0·76) compared with placebo. involving specific interactions of microbial structures www. CSF bactericidal activity has been shown in children who have meningitis due to cephalosporin-resistant pneumococci. partly because of our incomplete knowledge on the pathogenesis of neurological sequelae associated with bacterial meningitis.Review Panel: Empirical antimicrobial regimen for treatment of bacterial meningitis. and lower rates of severe hearing loss and long-term neurological sequelae.133. adjunctive treatment with dexamethasone was associated with lower case mortality. particularly for those infections in newborns. particularly in resource-limited settings.131 However.128 The beneficial effect of adjunctive dexamethasone treatment was evident in adults with bacterial meningitis. concomitant giving dexamethasone and vancomycin can reduce penetration of vancomycin into the CSF by virtue of the antiinflammatory activity of dexamethasone. wide availability. A recent multicentre. up to a maximum of 4 g daily) plus vancomycin (15 mg/kg every 6 h. fever curve. cephalosporinresistant pneumococcus) or organisms that are difficult to treat (eg. and because of its safety. low cost. or vancomycin (15 mg/kg every 6 h) can be added in the setting of suspected pneumococcal meningitis (eg. up to a maximum of 12 g daily) or ceftriaxone (50 mg/kg every 12 h. New information available on the pathogenesis of meningitis is likely to be useful for the prevention and treatment of bacterial meningitis. Monitoring of the clinical response (eg. double-blind randomised study in six Latin American countries showed that adjunctive treatment with oral glycerol (1·5 g/kg every 6 h for 48 h) prevents severe neurological sequelae in childhood meningitis (odds ratio 0·31. resulting in 38 treatment failure.com/infection Vol 10 January 2010 . resolution of symptoms and signs) and bacteriological responses to antimicrobial treatment. and oral administration. or cefotaxime (50 mg/kg every 6–8 h) can be used in the setting of suspected Gram-negative bacilli 1–3 months Ampicillin (50–100 mg/kg every 6 h) plus cefotaxime (75 mg/kg every 6–8 h) or ceftriaxone (50 mg/kg every 12 h).135 Glycerol is a hyperosmolar agent. For example. Future challenges Bacterial meningitis continues to be an important cause of mortality and morbidity throughout the world. effective strategies to prevent morbidity are lacking at present. Bacterial invasion and transcytosis in transfected human brain microvascular endothelial cells. Tuomanen EI. Berman PH. Gauczynski S. 2000. Cabanes D. Rubin LL. Stins M. Cebria A. Weiser JN. Blockade or inhibition of such host receptors or signalling molecules is efficient in preventing microbial traversal of the blood–brain barrier. Ring A. Changing epidemiology of pneumococcal meningitis after the introduction of pneumococcal conjugate vaccine in the United States. N Engl J Med 2009. Rubens CE. Badger JL. The role of humoral antibodies. Progress toward elimination of Haemophilus influenza type b invasive disease among infants and children—United States. Artenstein MS. Fu Q. 14 15 16 17 18 19 20 with the host receptors. Kondig JP. J Immunol 1933. Pelyola H. Annu Rev Neurosci 1999. Haik S. Jonas M. References 1 Klinger G. 102: 1087–97. “pathogenesis of bacterial meningitis”. Kim KS. Ammann AJ. Harvey D. 65: 5074–81. 30: 19–28. 88: F179–84. Invasion of brain microvascular endothelial cells by group B streptococci. 35: 35–42. Latsch K. Long-term outcome of neonatal meningitis. Long-term protection in children with meningococcal C conjugate vaccination: lessons learned. Warren RL. EMBO J 2005. Kent A. 22: 11–28.thelancet. Human immunity to the meningococcus. Burke B. “treatment of bacterial meningitis”. Wara DW. Three-year multicenter surveillance of pneumococcal meningitis in children: clinical characteristics. Hadler J. 6 Arditi M. et al. 6: 625–34. 348: 1737–46. Doran KS. Sadoff J. Influence of admission findings on death and neurological outcome from childhood bacterial meningitis. Vip. with the following search terms (alone and in combination): “neonatal bacterial meningitis”. 5 Roine I. Huang SH. 1998–2000. 3 Chang CJ. Molecular analysis of a novel bi-directional pathway. N Engl J Med 1976. 90: 897–905. Lecuit M. A putative receptor for Venezuelan equine encephalitis virus from mosquito cells. Nat Rev Neurosci 2003. 70: 5592–99. Cryz SJ Jr. Blood–brain barrier invasion by group B streptococcus depends upon proper cell-surface anchoring of lipoteichoic acid. with the emphasis on new information reported since 2000. Conflicts of interest I declare that I have no conflicts of interest. Mechanisms of microbial traversal of the blood–brain barrier. J Clin Invest 2009. 280: 1360–68. Clin Infect Dis 2008. Ferrieri P. Whitney CG. J Virol 1996. J Virol 2004. 115: 2499–507. Serotype-specific entry of dengue virus into liver cells: identification of the 37-kilodalton/67-kilodalton high-affinity laminin receptor as a dengue virus serotype 1 receptor. Engelson EJ. 61: 974–80. 13 Peltola H. 102: 347–60. 13: 302–17. www. Lameris-Martin NB. Sousa S. Cross AS. 360: 244–56. Huang LT. Eames M. Chang WN. Kim KS. Kim KS. Nguyen D. Brain Dev 2004. Influenzal meningitis: the relation of age incidence to the bactericidal power of blood against the causal organism. E coli invasion of brain microvascular endothelial cells in vitro and in vivo: molecular cloning and characterization of E coli invasion gene ibe10. Holt D. Correlation of maternal antibody deficiency with susceptibility to neonatal group B streptococcal infection. et al. Pneumococcal trafficking across the blood–brain barrier. Halket S. 119: 1638–40. et al. Decline in invasive pneumococcal disease after the introduction of proteinpolysaccharide conjugate vaccine. Only papers published in English were considered. J Biol Chem 2005. J Clin Invest 1998. Clin Microbiol Rev 2000. 46: 933–46.com/infection Vol 10 January 2010 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 Centers for Disease Control and Prevention. Thepparit C. 2 Stevens JP. The 37-kDa/67-kDa laminin receptor acts as the cell-surface receptor for the cellular prion protein. J Clin Invest 1992. N Engl J Med 2003. Fibronectin mediates Opc-dependent internalization of Neisseria meningitidis in human brain microvascular endothelial cells. Mahdavi J. The cell biology of the blood–brain barrier. 172: 178–86. Itabashi H. Gotschlich EC. J Exp Med 1969. Production of bacteremia and meningitis in infant rats with group B streptococcal serotypes. Orihuela CJ. et al. Infect Immun 1995. Beyene J. Chin CN. Predicting the outcome of neonatal bacterial meningitis. Fernandez J. Effect of pneumococcal conjugate vaccine on pneumococcal meningitis. Miller E. Smith JF. and eliciting host signalling molecules. 5: 851–57. Bacterial meningitis in infants: the epidemiology. Kasper DL. Pediatrics 2000. Effect of antibody concentration on opsonic requirements for phagocytosis in vitro of Streptococcus pneumoniae types 7 and 19. Pediatrics 1966. Borrow WR. Ludwig GV. “microbial invasion and/or traversal of the blood– brain barrier”. Mason EO Jr. to June. Kim KS. 46: 1664–72. Kim KS. 67-kDa laminin receptor promotes internalization of cytotoxic necrotizing factor 1-expressing Escherichia coli K1 into human brain microvascular endothelial cells. Outer membrane protein A and cytotoxic necrotizing factor-1 use diverse signaling mechanisms for Escherichia coli K1 invasion of human brain microvascular endothelial cells. Eur J Pediatr 2004. 46: 1248–52. Infect Immun 1993. 9 Chudwin DS. Wass CA. 7: 730–34. Khosravi A. 39 . 294: 753–56. et al. 51: 234–37. Chung JW. 63: 4470–75. Infect Immun 1980. Infect Immun 1997. Estimation of protective levels of anti-O-specific lipopolysaccharide immunoglobulin G antibody against experimental Escherichia coli infection. Kim KS. Staddon JM. Expert Rev Vaccines 2006. Microb Pathog 2001. Thornton J. Chung JW. Garcia-del Portillo F. Neonatal meningitis in England and Wales: sequelae at 5 years of age. Harvey D. J Clin Invest 2005. et al. Kim KJ. The K1 capsule is the critical determinant in the development of Escherichia coli meningitis in the rat. Neonatal meningitis. Cross AS. Prasadarao NV. Stins MF. 20: 5863–75. I. and “adjunct therapy of bacterial meningitis”. et al. 7 Kim KS. 2009 (date limits January. Moore MR. which formed the foundation for subsequent studies. Laminin receptor initiates bacterial contact with the blood brain barrier in experimental meningitis models. Nelson J.Review Search strategy and selection criteria The information for this Review was identified by searches of Medline in June. 27: 1023–32. Unkmeir A. Hsu HE. “diagnosis of bacterial meningitis”. Arch Dis Child Fetal Neonatal Ed 2003. Wass CA. Wright J. Smith DR. 38: 6–24. Grijalva CG. Farley MM. 10 Goldschneider I. et al. EMBO J 2001. A clinical and pathological study of 29 cases. Pekka Nuorti J. 11 Baker CJ. et al. et al. Mol Microbiol 2002. clinical features. Banker BQ. Halkets S. and this host-based approach presents a new approach in our strategies to prevent and treat bacterial meningitis. 106: 477–82. “bacterial meningitis in infants and children”. and prognostic factors. 12 Schiff DE. Peyrin JM. Stins M. Pathogenesis of bacterial meningitis: from bacteremia to neuronal injury. Shin S. Worldwide Haemophilus influenzae type b diseases at the beginning of the 21st century: global analysis of the disease burden 25 years after the use of the polysaccharide vaccine and a decade after the advent of conjugates. Shutt KA. 24: 2827–838. Proc Soc Exp Biol Med 1983. Nizet V. and outcome related to penicillin susceptibility and dexamethasone use. 26: 168–75. Nat Rev 2008. Griffin MR. 2009). Kim KS. 4: 376–85. Cossart P. 78: 12647–56. Pediatrics 1998. 4 de Louvois J. Tsai CJ. Microb Pathog 2003. Gemski P. MMWR Morb Mortal Wkly Rep 2002. 24: 273–84. Khan NA. gp96 is a receptor for a novel Listeria monocytogenes virulence factor. Earlier original articles were also included. Bradley JS. Perlman M. Clin Infect Dis 2008. Detrich G. 8 Fothergill LD. a surface protein. 129: 1307–26. and reasons for consulting a physician. Clin Microbiol Rev 1992. Clin Microbiol Rev 2004. Spellerberg B. Lu G. Schlaepfer DD. Wang Y. 73: 2923–31. 17: 260–65. Infect Immun 2000. 330: 1199–204. 268: 8053–59. 3: 1271–80. Fried D. et al. 6: 153–66. EMBO J 2000. Leenen PJ. et al. 5: 130–45. Tenenbaum T. Orihuela CJ. Phosphatidylinositol 3-kinase activation and interaction with focal adhesion kinase in E coli K1 invasion of human brain microvascular endothelial cells. 278: 16857–62. 74: 3967–74. Paul-Satyaseela M. Lumbar puncture in pediatric bacterial meningitis: defining the time interval for recovery of cerebrospinal fluid pathogens after parenteral antibiotic pretreatment. Bradley JS. Escherichia coli K1 induces IL-8 expression in human brain microvascular endothelial cells. Escherichia coli K1 RS218 interacts with human brain microvascular endothelial cells via type 1 fimbria phase-on bacteria. 74: 1360–67.com/infection Vol 10 January 2010 . Concentrated Gramstained smears prepared with a cytospin centrifuge. a C1q-binding protein is a receptor for the InlB invasion protein of Listeria monocytogenes. Wass C. Treatment strategies for central nervous system infections. Diseases that mimic meningitis: analysis of 650 lumbar punctures. Le Monnier A. 17: 161–64. Microbes Infect 2007. Vogel M. Soliemanzadeh P. Heath PT. Jager V. Murray PJ. Peterson LR. Kim KS. Nemani PV. 301: 373–75. Dryja D. La Scolea LJ Jr. EI. Ruuskanen O. Infect Immun 1999. Kim KS. 258–61. Improving the outcome of neonatal meningitis. Chung JW. Escherichia coli outer membrane protein A adheres to human brain microvascular endothelial cells. Bergman P. J Infect Dis 2001. Streptococcus pneumoniae anchor to activated human cells by the receptor for platelet-activating factor. Idanpaan-Heikkila I. 277: 15607–12. 16: 1052–56. et al. Lambert-Zechovsky N. Levy M. 7: 167–80. Tuomanen EI. Greiffenberg L. 345: 538–42. Cell 2000. Schaper PJ. Involvement of focal adhesion kinases in Escherichia coli invasion of human brain microvascular endothelial cells. et al. Pediatrics 2001. Kim KS. Cai M. Infect Immun 2005. McGowan KL. RhoA and Rac1 contribute to type III group B streptococcal invasion of human brain microvascular endothelial cells. Hensler M. 22: 229–34. Infect Immun 1999. Clin Pediatr 1990. Khan NA. Kim KS. Reinscheid DJ. 66: 5260–67. Datta K. Jonsson AB. Childhood bacterial meningitis: initial symptoms and signs related to age. Khan NA. et al. J Bacteriol 2007. 9: 1408–12. Theunissen CC. et al. Differential role of cytosolic phospholipase A2 in the invasion of brain microvascular endothelial cells by Escherichia coli and Listeria monocytogenes. Gray LD. Biochem Biophys Res Commun 2006. Shin S. Reddy MA. Stephens DS. Infect Immun 2005. Chen YH. Peltola H. Kim KS. Schroten H. Wong E. Hafiz A. Kim KJ. 9: 169–78. Yant SR. J Biol Chem 2003. 17: 323–47. Tenenbaum TT. Santoro F. Adherence to and invasion of human brain microvascular endothelial cells are promoted by fibrinogen-binding protein FbsA of Streptococcus agalactiae. InlB-dependent internalization of Listeria is mediated by the Met receptor tyrosine kinase. Plasmodium falciparum uses gC1qR/HABP1/p32 as a receptor to bind to vascular endothelium and for platelet-mediated clumping. Reddy MA. 9: 278–81. Central nervous system infection with Listeria monocytogenes. Gerard C. Massia SP. J Virol 2000. Kim KS. Goebel W. coli K1 invasion of brain microvascular endothelial cells. Hohmann EL. long-term intracellular growth and spread from macrophages to endothelial cells. Wass CA. Insinga A. www. Curr Opin Infect Dis 2009. Eur Cytokine Netw 2006. 67: 2103–09. Pandey K. Kim KS. Nemani PV. Kanegaye JT. Infect Immun 2005. Cell Microbiol 2007. ibeB. J Exp Med 1998. J Biol Chem 1993. Infect Immun 1998. 278: 25964–69. Grobbee DE. Bingen E. Lieber A. Cai M. Derksen-Lubsen G. Radin JN. Manchester M. Nizet V. Nature 1995. 103: 501–10. Oostenbrink R. Mylonakis E. Naujokas M. Covalently immobilized laminin peptide Tyr-Ile-Gly-Ser-Arg (YIGSR) supports cell spreading and co-localization of the 67-kilodalton laminin receptor with alphaactinin and vinculin. PLoS Pathogens 2007. Hong SJ. Valsamakis A. Schroten H. Banjerjee B. J Virol 2006. Greenfield RA. Structure of the human receptor tyrosine kinase Met in complex with the Listeria invasion protein InlB. Ghebrehiwet B. Tuomanen. 184: 732–37. 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 Niemann HH. Guglielmo C. FimH-mediated Escherichia coli K1 invasion of human brain microvascular endothelial cells. Valmari P. gC1q-R/p32. Park M.thelancet. Butler PJ. Cell Microbiol 2005. 67: 4751–56. Wass CA. J Biol Chem 2003. The gene locus yijP contributes to E. Zughaier S. 33 years’ experience at a general hospital and review of 776 episodes from the literature. Grimm D. 189: 1464–67. J Biol Chem 2002. Kim KS. Interaction of Listeria monocytogenes with human brain microvascular endothelial cells: InlB-dependent invasion. 80: 9831–36. required for penetration of brain microvascular endothelia cells. Medicine 1998. Doran KS. Drevets DA. Chung JW. Murti G. Shin S. Biswas AK. Hubbell JA. Kim KS. Cell Microbiol 2004. Science 2003. Galanakis E. Infect Immun 2006. Bloier C. Johansson L. Plant L. 37 kDa laminin receptor precursor modulates cytotoxic necrotizing factor 1-mediated RhoA activation and bacterial uptake. Moons KG. 73: 7827–35. Shen Y. Invasion of the central nervous system by intracellular bacteria. Signs of meningeal irritation at the emergency department: how often bacterial meningitis? Pediatr Emerg Care 2001. Martin A. Xu H. J Clin Microbiol 1984. et al. Asatryan L. 3. Pab N. Cytotoxic necrotizing factor 1 contributes to Escherichia coli K1 invasion of the central nervous system. Sokolova O. Wang Y. CD46 is a cellular receptor for group B adenoviruses. Adam R. Kim KS. Rao SS. Huang SH. 377: 435–38. Calderwood SB. Bacterial counts in cerebrospinal fluid of children with meningitis. 187: 631–40. Interaction of glycoprotein H of human herpesvirus 6 with the cellular receptor CD46. Shayakhmetov DM. Richards J. Greenstone HL. Eto DS.and tyrosine kinases in invasion and inflammatory cytokine release. Wass CA. 2. 73: 4404–09. Cell 2007. Gerard NP. 29: 254–55. et al. Nat Med 2003. Di Cello F. Kim KS. Cossart P. 130: 235–46. and 9. Biochem Biophys Res Commun 2005. 275: 36769–74. Maisey HC. Huang SH. Group B streptococcal pilus proteins contribute to adherence to and invasion of brain microvascular endothelial cells. Weiser JN. CD46 in meningococcal disease. Expert Opin Pharmacother 2009. Heppel N. Kim KJ. Mariani-Kurkdjian P. Kuhn M. Chitnis CE. 108: 1169–74. Ireton K. 146: 515–18. et al. Korvenranta H. Musher D. Fedorko DP. J Clin Microbiol 1982. Identification and characterization of an Escherichia coli invasion gene locus. et al. Streptococcus agalactiae invasion of human brain microvascular endothelial cells is promoted by the laminin-binding protein Lmb. Kay MA. 68: 6423–30. Clinical isolates of measles virus use CD46 as a cellular receptor. Interaction of Neisseria meningitidis with human brain microvascular endothelial cells: role of MAP. Join-Lambert OF. Exine S. Kim KS. Gaggar A.Review 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 40 Akache B. Rytkonen A. Cundell DR. Shanholtzer CJ. Reddy MA. Kim Y. Jägerhuber R. 77: 313–36. The 37/67-kilodalton laminin receptor is a receptor for adeno-associated virus serotypes 8. Teng CH. Moll HA. Adam R. Das A. 9: 714–20. et al. Quantitation of bacteria in cerebrospinal fluid and blood of children with meningitis and its diagnostic significance. Listeria monocytogenes-infected bone marrow myeloid cells promote bacterial invasion of the central nervous system. Lipooligosaccharide structure contributes to multiple steps in the virulence of Neisseria meningitidis. 19: 187–90. Sundqvist J. Lovkvist L. Eur J Clin Microbiol Infect Dis 1990. Shin S. Laboratory diagnosis of bacterial meningitis. 10: 1307–17. Fu Q. 19: 1458–66. J Biol Chem 2000. Galiza EP. Braun L. Shin S. β-Arrestin 1 participates in platelet-activating factor receptor-mediated endocytosis of Streptococcus pneumoniae. Eur J Pediatr 1987. Phosphorylcholine on the lipopolysaccharide of Haemophilus influenzae contributes to persistence in the respiratory tract and sensitivity to serum killing mediated by C-reactive protein. Lutsari I. Rapid diagnosis of bacterial meningitis using a microarray. 96 Tunkel AR. Basel: S Karger AG. Cottagnoud P. et al. Kennedy PG. J Formos Med Assoc 2008. Kravcik S. Osona B. 123 Grandgirard D.thelancet. Pedruzzi P. Antimicrob Agents Chemother 2008. Munoz C. Pharmacodynamics of gatifloxacin in cerebrospinal fluid in experimental cephalosporinresistant pneumococcal meningitis. et al. Levy C. 18: 188–93. et al. 335: 732–33. Murayama SY. J Clin Microbiol 2006. Stuertz K. Wang Q. 83 Corless CE. 76: 22–76. Presentation. Antimicrob Agents Chemother 2001. 89: 661–65. 21: 387–92. and Streptococcus pneumonia in suspected cases of meningitis and septicemia using real-time PCR. Schurch C. Ghaffar F. and mortality of patients with bacterial meningitis at an urban county medical center. Yamashita Y. a major cause of neonatal meningitis. 85 Lu JJ. de Boer RF. 107 McGeary SA. van Zwet AA. Li Y. Hsieh Y. 86 Wu Y-D. 117 Cottagnoud P. Kuppermann N. Wubbel L. Wolff M. Acosta F. Van Sorge NM. 1985: 83–89. against penicillin-sensitive or -resistant pneumococci in experimental meningitis. J Clin Microbiol 2005. 116 Gerber CM. 46: 2613–19. Saavedra J. Biros M. Wan CC. Antimicrobial agents in the treatment of bacterial meningitis. EFNS guideline on the management of community-acquired bacterial meningitis: report of an EFNS task force on acute bacterial meningitis in older children and adults. Frey J. Lu J. Clinical prediction rule for identifying children with cerebrospinal fluid pleocytosis at very low risk of bacterial meningitis. Eur J Clin Microbiol Infect Dis 1999. 39: 1267–84. Scand J Infect Dis 1986. et al. Meropenem alone and in combination with vancomycin in experimental meningitis caused by a penicillin-resistant pneumococcal strain. Shang S-Q. Craig WA. J Clin Microbiol 2008. Infect Dis Clin North Am 2004. Lee SY. Darmstadt GL. Perng CL. In: Christensen KK. Treatment failure with use of a third-generation cephalosporin for penicillin-resistant pneumococcal meningitis: case report and review. 89 Gill P. De La Rocque F. et al. 82 Chiba N. 41 . Cottagnoud M. Tunkel AR. Jafri H. 18: 866–70. Morozumi M. 107: 448–53. Antimicrob Agents Chemother 2007. 119 Schmidt H. Chan J. Tenover FC. 24: 1093–98. 10: 693–96. Neisseria meningitidis: evolution of penicillin resistance and phenotype in a children’s hospital in Barcelona. 18: 581–602. www. et al. Southern KW. Ghaemi A. Antimicrob Agents Chemother 1998. Tauber MG. Tsuda H. Kaplan SL. 115 Hedberg ST. 152: 378–82. J Clin Microbiol 2001. Wellmer A. Treatment failure in meningococcal meningitis. Dalhoff A. 105 John CC. JAMA 2007. 34: 2758–65. Friedland IR. Quagliarello VJ. BMS-284756 in experimental cephalosporin-resistant pneumococcal meningitis. Gonzalez-Cuevas A. PLoS ONE 2008. Antimicrob Agents Chemother 2003. Caugant DA. 87 Saha SK. Antimicrob Agents Chemother 2009. 93 Saha SK. Gemifloxacin is effective in experimental pneumococcal meningitis. 99 Miner JR. et al. Delays in the administration of antibiotics are associated with mortality from adult acute bacterial meningitis. 38: 2076–80. Potschka H. 95 Dubos F. Baqui AH. et al. 15: 649–59. Gene A. 53: 1561–66. 88 Seki M. Meningococcal meningitis during penicillin therapy for meningococcemia. First molecular characterization of group B streptococci with reduced penicillin susceptibility. et al. Sato S. Practice guidelines for the management of bacterial meningitis. Ward JI. 129: 862–69. 94 Nigrovic LE. 2000 to 2006: phenotypic and genotypic perspectives. 114 Jackson LA. J Emerg Med 2001. Pharmacokinetics and pharmacodynamics of antibiotics in meningitis. 106 Andes DR. Prospective study of use of PCR amplification and sequencing of 16S ribosomal DNA from cerebrospinal fluid for diagnosis of bacterial meningitis in a clinical setting. Activities of ertapenem. Wachino JI. time to antibiotics. Antimicrob Agents Chemother 1998. 39: 1553–58. Hartman BJ. 51: 2173–78. 49: 327–30. Clin Infect Dis 1997. 1991. Prevalence of Neisseria meningitidis relatively resistant to penicillin in the United States. Frechette D. Role of drug efflux transporters in the brain for drug disposition and treatment of brain diseases. J Antimicrob Chemother 2002. Neftel KA. Rapid detection of eight causative pathogens for the diagnosis of bacterial meningitis by real-time PCR. 44: 1447–52. Nau R. 103 Löscher W. Feng N. et al. 15: 92–98. J Clin Microbiol 2004. 122 Rodriguez-Cerrato V. 110 Dahesh S. Haemophillus influenza.com/infection Vol 10 January 2010 102 Sinner SW. Detrimental role of delayed antibiotic administration and penicillin-nonsusceptible strains in adult intensive care unit patients with pneumococcal meningitis: the PNEUMOREA prospective multicenter study. 100 Proulx N. J Clin Microbiol 2005.Review 81 Suwanangool S. Acosta F. 111 Turner PC. Guiver M. 113 Latorre C. 3: e3576. 97 Chaudhuri A. Kim KS. Eisenach KD. 13: 595–618. 27: 224–43. Juncosa T. Chen L-H. a new long-acting carbapenem. 42: 2650–55. Crit Care Med 2006. 43: 1581–86. Pfister M. Olcen P. Domingo P. Fox AJ. Kooistra-Smid AM. 52: 2915–18. Martinez-Martin P. Use of a serotype-specific DNA microarray for identification of group B streptococcus. 104 Ahmed A. Cottagnoud M. Gerber J. et al. Nicolas P. Clin Infect Dis 1994. Du L-Z. Antimicrob Agents Chemother 2008. Pediatr Infect Dis J 2005. Susceptibility of group B streptococcus to beta-lactam antibiotics. Maeno M. 112 Casado-Flores J. Antimicrob Agents Chemother 2000. Nucleosides Nucleotides Nucleic Acids 2008. et al. 52: 2890–97. 120 Lutsari I. Use of PCR with universal primers and restriction endonuclease digestions for detection and identification of common bacterial pathogens in cerebrospinal fluid. J Pediatr 2008. Clin Infect Dis 2004. Antimicrob Agents Chemother 1999. Sensitivity of the bacterial meningitis score in 889 children with bacterial meningitis. et al. Spain. Kung S. Cottagnoud P. Infect Dis Clin North Am 1999. et al. Kaczmarski EB. Chang F. 25: 1479. Q JM 2005. Leib SL. Tauber MG. 98: 291–98. Baker C. Charpentier J. 90 Ben R. J Clin Microbiol 2000. Tauber MG. Antibiotics and chemotherapy: neonatal group B streptococcal infections. 43: 1024–31. Hensler ME. Antibiotic susceptibility and characteristics of Neisseria meningitidis isolates from the African meningitis belt. Torigoe H. Rapid diagnosis of pneumococcal meningitis: implications for treatment and measuring disease burden. Lou J-T. Edwards-Jones V. 43: 876–81. Spencer NJB. Drugs Exp Clin Res 1983. Lancet 1990. Cottagnoud M. Fredlund H. 84 Schuurman T. Eng RH. J Infect Dis 1994. Pullen H. Heegaard W. 42: 1397–407. Christensen P. Smith SM. Macias CG. Ferrieri P. 109 Kimura K. Identification of serotype in culture negative pneumococcal meningitis using sequential multiplex PCR: implication for surveillance and vaccine design. Loopmediated isothermal amplification method targeting the lytA gene for detection of Streptococcus pneumoniae. Henne S. 47: 1943–47. Prevention of brain injury by the nonbacteriolytic antibiotic daptomycin in experimental pneumococcal meningitis. Ann Intern Med 1998. et al. 121 Smirnov A. 45: 3098–103. Wu X-J. Simultaneous detection of Neisseria meningitidis. 98 Aronin SI. Cefepime is efficacious against penicillin. 18: 347–52. Yamanaka N. Toye B. Pharmacodynamics of vancomycin for the treatment of experimental penicillin-and cephalosporinresistant pneumococcal meningitis. Community-acquired meningitis: risk stratification for adverse outcome and effect of antibiotic timing. Moxifloxacin in the therapy of experimental pneumococcal meningitis. et al. Antimicrobial susceptibility of group B streptococci. Prog Neurobiol 2005. 91 Wu L. 42: 734–40. 169: 438–41. 44: 767–70. Point mutation in the group B streptococcal pbp2x gene conferring decreased susceptibility to β-lactam antibiotics. Maier K. Gram stain-specific-probe-based real-time PCR for diagnosis and discrimination of bacterial neonatal sepsis. 92 Korczak B. Detection of bacterial antigens in body fluids by the Phadebact system. Suzuki S.and quinolone-resistant pneumococci in experimental meningitis. eds. J Infect Chemother 2009. Darmstadt GL. Use of diagnostic microarrays for determination of virulence gene patterns of Escherichia coli K1. Unemo M. Barquet N. Borrow R. 118 Cottagnoud P. Mapes A. 101 Auburtin M. Acta Paediatr 2000. Schrenzel J. Nucleic acid isothermal amplification technologies: a review. et al. 297: 52–60. Eur J Neurol 2008. 108 Kim KS. McIntyre P. Backer V. In: Pickering LK. Elk Grove Village. 34: 227–35. J Pediatr 1981. Association between persistence of pneumococcal meningitis and dexamethasone administration. McCoig CC. 135 Peltola H.thelancet. 125 Grady RW. Red book: 2009 report of the Committee on Infectious Diseases. 127 Andersen J. 54: 353–57. (1): CD004405. 28th edn. Tauber MG. 1991. MMWR Morb Mortal Wkly Rep 1996. 99: 924–26. www. Wandall JH. Adjuvant glycerol and/or dexamethasone to improve the outcomes of childhood bacterial meningitis: a prospective. Georgia. 128 Van De Beek D. 129 McIntyre PB. placebocontrolled trial. Clin Infect Dis 2007. Taber LH. Gerber J. Atlanta. JAMA 1997. 2009: 524–35. Garenoxacin (BMS-284756) and moxifloxacin in experimental meningitis caused by vancomycin-tolerant pneumococci. Postnatal causes of developmental disabilities in children aged 3–10 years. 45: 130–34. 60: 210–15.com/infection Vol 10 January 2010 . ed. Shinhoj P. et al. Hanssen M. 133 Leib SL. Pediatr Res 2006. randomized. 134 Spreer A. 132 Klugman KP. Acute meningococcal meningitis: analysis of features of the disease according to the age of 255 patients. 278: 925–31. Pediatr Res 2003. Saavedra J. Corticosteroids for acute bacterial meningitis. IL: American Academy of Pediatrics. Bradley JS. 130 American Academy of Pediatrics. Antimicrob Agents Chemother 1995. Fernandez J. Kaplan SL. J Infect 1997.Review 124 Rodriguez-Cerrato V. Bifrare YD. De Gans J. Friedland IR. 45: 1277–86. Loeffler JM. Dexamethasone increases hippocampal neuronal apoptosis in a rabbit model of Escherichia coli meningitis. Dexamethasone as adjunctive therapy in bacterial meningitis. Pneumococcal infections. Antimicrob Agents Chemother 2003. Berkey CS. Heimgartner C. Cochrane Database Syst Rev 2007. 47: 211–15. 42 131 Brady MT. Bactericidal activity against cephalosporin-resistant Streptococcus pneumoniae in cerebrospinal fluid of children with acute bacterial meningitis. double-blind. 126 Centers for Disease Control and Prevention. Prasad K. et al. 4: 523–630. et al. Corticosteroids for acute bacterial meningitis. Dexamethasone aggravates hippocampal apoptosis and learning deficiency in pneumococcal meningitis in infant rats. et al. A meta-analysis of randomized clinical trials since 1988. Voldsgaard P. Systemic quinolone antibiotics in children: a review of the use and safety. Roine I. 39: 1988–92. King SM. Expert Opin Drug Saf 2005.
Copyright © 2024 DOKUMEN.SITE Inc.